ArticlePDF AvailableLiterature Review

Superficial Bladder Cancer Therapy

Wiley
The Scientific World Journal
Authors:

Abstract and Figures

Bladder cancer treatment remains a challenge despite significant improvements in preventing disease progression and improving survival. Intravesical therapy has been used in the management of superficial transitional cell carcinoma (TCC) of the urinary bladder (i.e. Ta, T1, and carcinoma in situ) with specific objectives which include treating existing or residual tumor, preventing recurrence of tumor, preventing disease progression, and prolonging survival. The initial clinical stage and grade remain the main determinant factors in survival regardless of the treatment. Prostatic urethral mucosal involvement with bladder cancer can be effectively treated with Bacillus Calmette-Guerin (BCG) intravesical immunotherapy. Intravesical chemotherapy reduces short-term tumor recurrence by about 20%, and long-term recurrence by about 7%, but has not reduced progression or mortality. Presently, BCG immunotherapy remains the most effective treatment and prophylaxis for TCC (Ta, T1, CIS) and reduces tumor recurrence, disease progression, and mortality. Interferons, Keyhole-limpet hemocyanin (KLH), bropirimine and Photofrin-Photodynamic Therapy (PDT) are under investigation in the management of TCC and early results are encouraging. This review highlights and summarizes the recent advances in therapy for superficial TCC.
Content may be subject to copyright.
Original Articles
TheScientificWorldJOURNAL (2004) 4 (S1), 387–399
ISSN 1537-744X; DOI 10.1100/tsw.2004.81
Superficial Bladder Cancer Therapy
Emmanuel Schenkman, M.D. and Donald L. Lamm, M.D.
al
West Virginia University, Morgantown, WV
Previously published in the Digital Urology Journ
Bladder cancer treatment remains a challenge despite significant improvements in
preventing disease progression and improving survival. Intravesical therapy has been
used in the management of superficial transitional cell carcinoma (TCC) of the urinary
bladder (i.e. Ta, T1, and carcinoma in situ) with specific objectives which include treating
existing or residual tumor, preventing recurrence of tumor, preventing disease
progression, and prolonging survival. The initial clinical stage and grade remain the main
determinant factors in survival regardless of the treatment. Prostatic urethral mucosal
involvement with bladder cancer can be effectively treated with Bacillus Calmette-Guerin
(BCG) intravesical immunotherapy. Intravesical chemotherapy reduces short-term tumor
recurrence by about 20%, and long-term recurrence by about 7%, but has not reduced
progression or mortality. Presently, BCG immunotherapy remains the most effective
treatment and prophylaxis for TCC (Ta, T1, CIS) and reduces tumor recurrence, disease
progression, and mortality. Interferons, Keyhole-limpet hemocyanin (KLH), bropirimine
and Photofrin-Photodynamic Therapy (PDT) are under investigation in the management
of TCC and early results are encouraging. This review highlights and summarizes the
recent advances in therapy for superficial TCC.
DOMAIN: urology
INTRODUCTION
ourth most common cancer among men and the eighth most common cancer among Bladder cancer is the f
women. It is estimated that 54,500 new cases of bladder cancer will be diagnosed in 1997 and 11,700
bladder cancer deaths will occur
1
. The incidence of bladder cancer has increased 36% in the United States
from 1956 to 1990 and mortality rates declined 8% between 1980-1995
2
. Intravesical immunotherapy has
had a positive impact on disease progression and has influenced the decline in mortality. A proper
understanding of the natural history of bladder cancer and significant prognostic factors is critical for
management of this disease. Seventy-four percent of cases are superficial at the time of diagnosis, of
which 70% are stage Ta and 30% are stage T1
3
. Low-grade non-invasive tumors may be treated with
resection and fulguration. However, despite complete tumor resection, two thirds of patients will develop
tumor recurrence in five years and by 15 years 88% of patients will develop a recurrence
4
. Progression
from superficial bladder cancer to deep muscle invasion occurs in 15% of patients
5,6
. The high rate of
tumor recurrence and potential progression provides an opportunity to institute chemoprevention or
prophylactic therapy. In this review we highlight the recent advances in intravesical therapy of bladder
and summarize the important role of intravesical immunotherapy in the management and prophylaxis of
superficial transitional cell carcinoma (TCC) of the urinary bladder.
387
Schenkman and Lamm: Superficial Bladder Cancer Therapy TheScientificWorldJOURNAL (2004) 4 (S1), 387–399
INDICATIONS FOR INTRAVESICAL THERAPY
Since its introduction in the late 1950ís, intravesical therapy has been used in the management of
superficial bladder cancer with three specific goals. These include eradicating existing/residual tumor,
or resection, and preventing progression of
disease. The objectives of intravesical therapy should be tailored to the patient. Intravesical therapy is
is
reco
Intravesical chemotherapy became popular in the 1960ís when thiotepa was shown to reduce tumor
recurrence and eliminate one third of papillary tumors
4
. Unlike systemic chemotherapy, responses to
ses are also
dependent on the duration of exposure which is short and limited by bladder capacity. Cytotoxic drugs are
show an advantage of chemotherapy in reduction of tumor
recu
preventing recurrence of tumor after complete bladder tum
most effective when tumor burden is minimized by transurethral resection of papillary disease and/or
fulguration of visible areas of carcinoma in situ (CIS). The suspected biologic behavior of the patient's
tumor remains an important determinant factor in the decision of intravesical therapy. Consequently, a
grade III tumor at high risk for recurrence and progression constitutes an accepted indication for
intravesical therapy. In the absence of other risk factors for progression, intravesical therapy is not
required for grade I/Ta (0) lesions which have a progression rate of only 2-4%
7
. However, multifocal Ta
disease with or without CIS is a relative indication for intravesical therapy
8
. Stage T1 disease, irrespective
of grade, has demonstrated the biological ability to invade, and has a reported progression rate of 29%
9
.
Intravesical therapy is therefore justified to prevent progression to muscle invasion. In his analysis of
prognostic factors in a cohort of 505 patients with TCC, Lipponen
10
reported that the initial clinical stage
and grade remain the main determinant of survival, irrespective of the treatment. CIS has a high risk of
disease progression, with an average 54% developing invasive disease at 5 years
11
. Therefore, the
presence of even small foci of CIS should be considered as a definite indication for intravesical therapy.
Intravesical immunotherapy is now the first line of treatment for diffuse CIS and has replaced cystectomy
as the initial therapy. Multifocal superficial disease irrespective of grade or stage is also associated with
increased risk of tumor recurrence and progression, and constitutes an indication for adjuvant intravesical
therapy. Other relative indications for intravesical therapy include low grade Ta disease recurring within 2
years, persistent positive urine cytology localized to bladder and urothelial dysplasia or severe atypia.
Prostatic urethral involvement with CIS carries a high risk of progression and poor prognosis and
should be treated aggressively. The use of intravesical Bacillus Calmette-Guerin (BCG) immunotherapy
has effectively spared cystectomy in many of these patients
12
. Intravesical chemotherapy appears to be
ineffective in the treatment of the prostatic urethra. Transurethral resection of the prostate
mmended for tumor staging and to open the bladder neck in order to allow BCG to bathe the prostatic
urethra. Reports of response to topical chemotherapy in muscle-invasive disease are lacking, therefore
such patients are not treated with intravesical therapy.
INTRAVESICAL CHEMOTHERAPY IN SUPERFICIAL BLADDER CANCER
topical chemotherapy are proportional to drug concentration rather than drug dose
13
. Respon
active against DNA in rapidly dividing cells.
In a review of over 4,000 patients enrolled in controlled intravesical trials, Traynelis and Lamm
14
reported that the average net benefit of intravesical chemotherapy over transurethral resection alone is a
modest 14% at 1 to 3 years. Of the 23 reported clinical trials, 13 demonstrated statistically significant
reduction in tumor recurrence. Most studies
rrence for the first two or three years. Melekos
15
reported that epirubicin prevented tumor recurrence
in 60% of treated patients versus 41% of controls during a mean follow-up of 32 months, which is
relatively a short-term follow-up. However, long term results with thiotepa, doxorubicin and mitomycin C
(MMC) demonstrated that the percent of patients suffering recurrence at 5 or more years is just as high, if
not higher, in patients receiving intravesical chemotherapy than in controls. Maintenance chemotherapy
has been shown to offer no advantage and perhaps even a disadvantage. Oosterlinck
16
reported recently a
reduction in tumor recurrence in patients with solitary Ta or T1 tumors treated with a single early
388
Schenkman and Lamm: Superficial Bladder Cancer Therapy TheScientificWorldJOURNAL (2004) 4 (S1), 387–399
postoperative instillation of epirubicin. In 399 patients, tumor recurrence was reduced from 41% in
controls to 29% with epirubicin (p=0.015). Intravesical chemotherapy in the absence of tumor cells
should not be beneficial since it acts directly on such cells. Therefore, the concept of preventing future
urothelial tumor recurrence with intravesical cytotoxic chemotherapy is illogical.
Although intravesical chemotherapy has demonstrated reduction in short-term tumor recurrence rates,
it has not altered disease progression. Progression data are available on more than 2,000 patients enrolled
in prospective controlled chemotherapy studies
14
. No statistically significant reduction is found in the risk
of disease progression with the use of thiotepa, doxorubicin, mitomycin, or epirubicin in those studies.
Mo
d in 13 out
of 3
dilution by constant production of
urin
BCG is currently the most effective intravesical agent for the treatment and prophylaxis of superficial
bladder cancer. BCG is recognized as a nonspecific immune stimulant. Intravesical BCG induces
ith infiltration of a broad range of cell types. BCG may activate
macrophages, T lymphocytes, B lymphocytes, natural killer cells (NK), and killer cells
20
. Intravesical
reover, the mean rate of progression for those treated with intravesical chemotherapy was 7.5%
compared with 6.9% for the control groups
14
. Similar results have been reported by the EORTC and MRC
meta-analysis of over 2500 patients. These investigators demonstrated that with a mean follow-up of 7
years, chemotherapy reduced long-term recurrence by 7%, but had no effect on progression
17
.
Intravesical chemotherapy can be beneficial in the management of CIS with reported complete
response rates range from 34 to 42%. Sekine et al.
18
have reported the results of mitomycin and
doxorubicin sequential therapy in 43 CIS patients. Thirty-two patients (74%) achieved complete response
(CR), but despite maintenance therapy, with either MMC or doxorubicin, recurrences occurre
2 (41%). Twenty-six of 32 (81%) complete responders remained disease free during a mean follow up
45 (range 10-84) months. Maintenance therapy failed to show any positive impact on recurrence rate.
Three of the 32 complete responders and 5 of 11 nonresponders suffered progression including invasive
cancers in four, metastatic disease in two and both conditions in two.
The reasons for the inability of intravesical chemotherapy to affect progression or enhance long-term
reduction in recurrence are under investigation. Wientjes et al.
19
reported that the putative variable and
inconsistent response of intravesical MMC might be due to physicochemical and hemodynamic factors
such as incomplete bladder emptying at treatment, low urine pH, or
e. Results of their ongoing Phase III clinical trial will indicate whether or not controlling these
variable improve cytotoxic efficacy of mitomycin .
MECHANISM OF ACTION
inflammation of the bladder w
BCG immunotherapy results in cytokine production, including interleukins 1(IL-1), 2(IL-2), and 6(IL-6),
interferon gamma, and tumor necrosis factor alpha (TNF-a)
21
, which can be measured in the urine for
many hours after instillation. McAveray et al.
22
reported that BCG induces a local Type II immunologic
response which may be mediated by Interleukin (IL) 4; IL-4, IL-10, the later cytokines may suppress cell-
mediated responses. These cytokines also cause a shift to Type I response with the subsequent
development of a protective antitumor response. Ratliff et al.
23
investigated the role of CD
4
and CD
8
lymphokines in the antitumor response of BCG and reported that there is no evidence of induction of
protective systemic immunity after BCG. However, they reported a requirement of T-lymphocytes, and
CD
4
and CD
8
subsets in BCG-mediated antitumor activity. They concluded that BCG-mediated antitumor
activity is a localized phenomenon. BCG stimulates cytokine production, and this in turn enhances NK
cell activity, which increases after BCG immunotherapy
24,25
. Conti et al.
26
reported that
immunotherapeutic effects of BCG in bladder cancer patients are related to its capacity to prime
macrophages that enhance the release of TNF-a and IL-I alpha, which are involved in tumor killing. BCG
produces a T-cell mediated immune response that has been linked to antitumor activity in both humans
and mice
27
. The antineoplastic effect of BCG is most likely the result of a combination of enhanced
activity of various arms of the immune system.
389
Schenkman and Lamm: Superficial Bladder Cancer Therapy TheScientificWorldJOURNAL (2004) 4 (S1), 387–399
After intravesical instillation, live mycobacteria attach to the urothelial lining, facilitated by
fibronectin, a component of the extracellular matrix
28
. Integrin is required for the direct attachment and
internalization of BCG by bladder tumor cells
28-30
. This process leaves bacterial cell surface glycoproteins
atta
To use immunotherapy effectively in the management of bladder cancer or other malignancy, it is
een immunotherapy and
chemotherapy. Currently chemotherapy is limited in specificity, and basically inhibits or destroys rapidly
x 1010) colony-forming units (CFU) are effective, but responses have been
rep
Long-term follow-up studies have consistently demonstrated prolonged protection from tumor recurrence
est that optimal BCG intravesical immunotherapy also
reduces tumor progression and mortality
. All six clinical studies comparing surgery alone with
ched to epithelial cell membranes, and this antigen is thought to mediate the immune response
31
.
Tumor cell motility is also thought to be inhibited by BCG through a mechanism involving the BCG-
fibronectin-tumor cell interaction
32
. Bladder biopsies following BCG administration show increased
expression of human leukocyte antigen (HLA)-Dr antigen on tumor cells and infiltration of tumor and
stroma with lymphocytes, predominantly T helper cells, and macrophages. The helper/suppressor ratio in
infiltrating lymphocytes is increased. Changes in peripheral blood are also seen, including heightened
immunoproliferative response to BCG antigen and production of specific antibody
33-34
.
PRINCIPLES OF BCG IMMUNOTHERAPY
important to consider basic principles and understand the differences betw
dividing cells. Generally, tumor cell destruction is proportional to drug concentration so treatments are
pushed to the limit of tolerance. In contrast, immunotherapy may be either nonspecific or specific. More
often than not, optimal responses to immunotherapy are seen at less than the maximum tolerated dose
because high doses invoke complex immune regulatory mechanisms. The typical dose response curve
with biological response modifiers such as BCG is therefore bell shaped with optimal response occurring
at intermediate doses
35
.
The optimal dose of BCG remains to be defined, and may, like the optimal treatment schedule, vary
from patient to patient. Current data suggest that intravesical doses between one hundred million (1 x
108) and one billion (1
orted with doses as low as 10 million CFU or 1 mg BCG
36
. The wide variation in effective clinical
doses probably relates to the mode of administration. In intravesical instillation, only those organisms that
attach to the bladder wall stimulate an immune response. Therefore, consideration must also be given to
avoid administration of medications that can limit the effectiveness of the dose given. Agents that inhibit
clot formation reduce fibronectin expression, which may reduce BCG attachment, immune stimulation,
and antitumor activity
37-39
. Similarly, concern has been raised that administration of antitubercular
antibiotics such as isoniazid (INH), which inhibit intravesical BCG attachment and immune stimulation in
the guinea pig model
40
, may also reduce the efficacy of BCG therapy. However, Stassar et al.
41
reported
that INH does not impair the local immunological stimulation after intravesical BCG. Until additional
data becomes available, INH, trimethoprim/sulfamethoxazole, and quinolones should be used with
caution in patients receiving BCG. However, these antibiotics should be used without hesitation to treat
the side effects of BCG or intercurrent infection.
EFFICACY OF BCG IMMUNOTHERAPY
by BCG
42-44
as well as increasing evidence to sugg
42-45
intravesical BCG immunotherapy demonstrated a highly significant advantage of BCG treatment
45-50
(Table 1). Direct randomized comparisons of BCG immunotherapy with intravesical chemotherapy have
390
Schenkman and Lamm: Superficial Bladder Cancer Therapy TheScientificWorldJOURNAL (2004) 4 (S1), 387–399
TABLE 1
Effect of Intravesical Bacillus Calmette-Guerin on Recurrence in Control Studies
CONTROL (TURBT) BACILLUS CALMETTE-
GUERIN
Authors and
References
Total
Number of
Patients
Number Recurred
#(%)
Number Recurred
#(%)
P Value
Lamm 57 27 14 (52%) 30 6 (20%) <0.001
Herr et al 86 43 41 (95%) 43 18 (42%) <0.001
Herr et al 49 26 26 (100%) 23 8 (35%) <0.001
Pagano et al 133 63 52 (83%) 70 18 (26%) <0.001
Melekos et al 94 32 19 (59%) 62 20 (32%) <0.02
Krege et al 224 122 56 (46%) 102 26 (26%) 0.003
TOTAL 643 313 208 (67%) 330 96 (24%)
KEY: TURBT = transurethral resection of bladder tumor
also demonstrated a statistically significant decrease in tumor recurrence rate with BCG compared with
thiotepa, doxorubicin, and mitomycin C (MMC)
12, 51-58
(Table 2). The Southwest Oncology Group
recently compared TICE BCG (50mg) and MMC (20mg) in 469 randomized high-risk patients with stage
Ta or T1 disease
57
. Both treatments were given weekly for six weeks then monthly for one year. A 20mg
dose of MMC was previously reported to be the optimum dose
58
. In the MMC arm, tumor reoccurred in
33% of patients with a median time to recurrence of 18.4 months. With a median follow-up of 30 months,
60% of patients in the BCG arm were without tumor recurrence as opposed to 46% of patients in the
MMC arm (p=0.017). No toxicity was seen in 18% of the BCG arm or in 30% of the MMC group
p<0.003). Melekos et al.
59
reported a recent series of 161 patients enrolled in a three-arm study of
intravesical prophylaxis with epirubicin versus BCG versus transurethral resection (TUR) alone. The
authors reported that 60% of epirubicin-treated patients, 68% of BCG-treated patients, and 41% of control
subjects remained free from recurrences at a median follow-up of 33 months. Epirubicin and BCG were
both superior to TUR alone; however, BCG was significantly better than epirubicin in preventing
recurrence of stage T1 and high-grade tumors. Cookson and Sarosdy
60
also demonstrated the
effectiveness of BCG in high-risk stage T1 patients in their trial; 91% of those treated with intravesical
BCG immunotherapy were free of disease at a mean follow-up of 59 months.
The effect of BCG on tumor progression has been investigated in three randomized studies, each of
which found a statistically significant reduction in progression to muscle invasion or metastasis
43,45,61
.
Lamm demonstrated a reduction in progression to muscle invasive disease in 8% of controls compared to
3% in the BCG group
45
. This positive impact on progression has resulted in improved survival. A
controlled trial from Memorial Sloan-Kettering showed persistent reduction in both tumor recurrence and
progression after ten years follow-up
62
. However, the reduction in tumor progression did not extend to
fifteen years. Overall, 53% of high-risk patients had progression with a disease-specific survival of 63%.
Thus, even after apparent successful treatment with BCG, patients remain at risk for progression,
recurrence, and mortality and require vigilant long-term surveillance. In another report, Herr et al.
61
reported that within a median follow-up of eight months, mortality rate was reduced from 32% in TUR
alone patients to 14% in BCG-treated patients (p<0.032). Herr and associates reported that BCG
improved a five-year survival to 87% versus 63% for TUR (p=0.016)
44
. This author has also reported that
391
Schenkman and Lamm: Superficial Bladder Cancer Therapy TheScientificWorldJOURNAL (2004) 4 (S1), 387–399
TABLE 2
Recurrence in Controlled Comparison Trials
Study/References BCG Thiotepa Doxorubicin Mitomycin C P Level
Brosman 0 47% <0.01
Rodriques Netto and Lemos 7% 43% <0.01
Martinez-Pineiro et al 17% 36% 43% <0.01
BCG/Thiotepa Average 7% 42%
Lamm et al 63% 83% <0.02
BCG/Doxorubicin Average 38% 63%
Debruyne et al 30% 25% NS
Juahianen et al 288% 62% <-0.01
Rubben et al 35% 35% NS
Witjes et al 29% (RIVM) 26% NS
34% (TICE) NS
Lamm et al 20% 33% <0.01
BCG/Mitomycin Average 29% 36%
BCG Average 25%
Chemotherapy Average 43%
KEY: BCG = bacillus Calmette-Guerin
cancer deaths were reduced from 37% to 12% (p<0.01) and that the cystectomy rate was reduced from
42% to 26% (p<0.0001) in patients with BCG
61
. Nadler et al.
43
demonstrated the durability of a single
course of BCG which kept 28% (29/104) of the patients tumor-free at 11 years. However, of the 66
patients who received a second six-week course of BCG for recurrent tumors after failing the intravesical
six-week course, 27(41%) remained tumor-free at 11 years. Witjes et al.
62
confirmed that effectiveness of
BCG in reducing tumor progression in high-risk patients who had failed prior intravesical chemotherapy
for recurrent superficial TCC.
BCG is also effective in the intravesical treatment of CIS. With over 1,000 patients from several
series, the average complete response rate of CIS to BCG is in excess of 70%
11
. By comparison, complete
response rates for chemotherapy average less than 50%, and in general, fewer than 20% of patients treated
with chemotherapy remain disease-free long-term
42
.
In contrast to intravesical chemotherapy, data suggest that maintenance therapy with BCG improves
long-term results. In a recent report by the Southwest Oncology Group
57
with optimal BCG
immunotherapy for recurrent superficial transitional cell carcinoma (CIS, Ta, T1), the complete response
(CR) rate was 87% and long-term disease-free status was maintained in 83% of patients. In CIS patients
treated with BCG, the complete response at six months post-therapy is increased from 73% to 87%
(p<.04) with three additional instillations given at six monthly intervals for maintenance
42
. Maintenance
BCG using three weekly instillations increased long-term disease-free status from the expected 65% to
83%. In patients with papillary TCC, maintenance BCG given in a series of three weekly treatments at
three months, six months, and every six months for three years, dramatically reduced tumor recurrence
(p<.0001) when compared with a single six-week course
63
. Long-term disease-free status was increased
from 50% in the induction-only group to 83% in the maintenance therapy group (p<0.000001). More
392
Schenkman and Lamm: Superficial Bladder Cancer Therapy TheScientificWorldJOURNAL (2004) 4 (S1), 387–399
importantly, this maintenance therapy has resulted in statistically significant improvement of patient
survival as compared to induction-only. In 391 randomized patients, the excellent 86% survival at four
years observed with induction therapy was improved to 92% in patients receiving maintenance BCG
(p<0.04)
63
. The current recommended maintenance BCG regimen ñ and the only regimen found to be
superior to a single, six-week induction ñ employs three weekly instillations of 105 to 108 CFU of
Connaught BCG, three months after initiation of treatment
63
. Three weekly instillations are repeated at six
monthly intervals for three years. The second or third weekly maintenance treatment is given only if the
preceding instillation was without increased side effects. Investigators have reported that in low-dose
BCG, 27mg/3x108 CFUs were efficacious, yielding a CR of 84%
48
, and some have seen a reduction in
toxicity.
It has been suggested that the efficacy of BCG can be improved further by high-dose vitamins
67
.
Lamm et al.
65
reported that daily high-dose vitamins A, B6, C, and E (Oncovite, Mission Pharmacal, San
Antonio, TX) versus recommended daily allowances (RDA) produced further protection from recurrence
in patients treated with BCG. The five-year estimates of tumor recurrence were 91% in the RDA group
and 41% in the megadose vitamin group. Overall recurrence was 24 of 30 (80%) patients in the RDA
group, and 14 of 35 (40%) in the high-dose arm
64
. Further research is needed to confirm this study and
identify which specific vitamins offer the protection from tumor recurrence. Other attempts to improve
BCG immunotherapy, such as the addition of intradermal BCG inoculation, have not yet been
successful
64
.
COMPLICATIONS OF BCG INTRAVESICAL THERAPY
Intravesical BCG presumably stimulates an immune response to the tumor and thus is associated with
unique side effects. Dysuria and urinary frequency are expected as a consequence of the inflammatory
response, and cystitis is the most frequent adverse reaction-occurring in up to 90% of cases
65,66
.
Hematuria may occur with cystitis and is seen in one-third of patients
66
. Irritative bladder symptoms are
unlikely in the week after the first intravesical BCG
66
. Side effects of BCG generally increase with
successive treatments, unless the dose of antibiotics is reduced or prophylactic antibiotics are given.
Patients with symptoms lasting more than 48 hours can be treated with 300mg INH daily
69
. This
treatment is continued only while the symptoms of hematuria and cystitis persist and is reinstituted one
day before subsequent BCG instillation and continued for three days. According to Stassar and
associates
41
, INH does not impair the local immunological stimulation after intravesical BCG or the
efficacy of BCG. BCG treatments are postponed until all side effects from previous instillations have
resolved. BCG is a live organism, and even though virulence has been dramatically attenuated, regional or
systemic infection may occur. BCG organisms usually are gone within a few days of instillation but have
been reported to persist in the urinary tract for at least 16.5 months after intravesical BCG
68
. Initial
estimates of the incidence of BCG sepsis were in the range of 0.04% and 10 patients died following
intravesical BCG
65
. The incidence of sepsis has dropped dramatically after the precaution of not
administering BCG after traumatic catheterization or in the presence of continued symptoms of BCG
infection. When BCG sepsis does occur, we now recommend INH 300mg, rifampin 600mg, and
prednisone 40mg daily. Prednisone is continued until sepsis abates and is then tapered gradually over the
next two to four weeks. Rifampin and INH are continued for three to six months, depending on the
severity and duration of the reaction. Animal studies
67
have confirmed that this regimen significantly
improves survival and no patient receiving this regimen has died of BCG sepsis. The diagnosis of BCG
sepsis is made by clinical presentation with high fever, shaking chills, and then hypotension. It is
important to proceed with antibiotic treatment without waiting for culture results when systemic BCG
infection is suspected. Typically, cultures are negative, even in the face of clinical sepsis. Molecular
techniques to identify BCG DNA may prove useful in the future
69
.
393
Schenkman and Lamm: Superficial Bladder Cancer Therapy TheScientificWorldJOURNAL (2004) 4 (S1), 387–399
OTHER IMMUNOTHERAPEUTIC AGENTS:
Interferons
Interferons (IFNs) are host-produced glycoproteins that act to mediate immune responses through
antiviral, anti-proliferative, and immunoregulatory activities. Torti et al.
70
reported 25% CR in patients
with recurrent papillary TCC, and 32% CR and 26% partial response (PR) ñ persistent positive cytology ñ
in 19 patients with refractory CIS treated with intravesical IFN-alpha. In a multicenter randomized study,
Glashan[71] found tumor response to INFa was dose dependent: there was a 5% CR observed with a dose
of 10 x 106 units, and a 43% CR with a dose of 100 x 106 units in patients treated for CIS. In our recent
review, IFN-alpha prophylaxis in TCC resulted in a freedom from recurrence in 21-62% of patients
within median follow-ups of 6-36 months (Riggs D, Nseyo UO, Lamm DL, unpublished observations).
Adverse reactions following intravesical IFN-alpha therapy are relatively mild and include flu-like
symptoms of fever, chills, fatigue, and myalgia, which occur in up to 27% of patients.
Keyhole Limpet Hemocyanin
Keyhole-limpet hemocyanin (KLH), a highly antigenic respiratory pigment of the mollusc Megathura
cranulata, is a nonspecific immune stimulator that also has been investigated as an intravesical agent.
Jurincic et al.
72
reported that KLH was better than MMC in prevention of superficial TCC recurrence.
Flamm et al.
73
compared KLH to ethoglucid for prophylaxis in patients who failed intravesical
chemotherapeutics agents, and reported no difference in efficacy. Lamm et al.
74
reported a CR in 25/51
(45%) patients and a PR in 12 (21%) patients with 2mg, 10mg, or 50mg of intravesical KLH for six
weeks. The best responders were patients with CIS: of 19 CIS patients, 11 (58%) had a CR. Ten (50%) of
20 patients with papillary TCC demonstrated a response, and 4 (33%) of the 12 patients with both forms
of bladder cancer showed response. Wishahi et al.
75
reported that KLH reduced tumor recurrence by 60%
in bilharzial bladder with papillary TCC. The advantage of KLH is its apparent lack of toxicity. Lamm et
al.
76
reported that crude preparation of KLH offered greater antitumor activity than the purified KLH
compound, although this has not been investigated in clinical trial.
Bropirimine
Bropirimine, an oral immunomodulator, has shown efficacy in therapy of bladder and upper tract TCC.
Bropirimine is an aryl pyrimidine with broad spectrum immunostimulatory activity. The spectrum of
activity includes induction of endogenous interferons, IL-I, and TNF. Bropirimine also stimulates B-cell
proliferation, NK cells, lymphokine-activated killer cells (LAK), and macrophage activity. Sarosdy et al.
77
reported that bropirimine induced a complete response in 27 of 52 (52%) patients treated for residual
disease. The best responders were patients without prior intravesical therapy. Of those 10 patients, seven
(70%) had a complete response. The median follow-up for the series was 12 months and toxicity was dose
related. In a separate investigation, Sarosdy et al.
78
reported the results of 25 patients with unilateral or
bilateral positive cytology with negative retrograde pyelography. Ten of 19 (53%) evaluable patients
showed negative cytology following oral bropirimine therapy. Four patients showed a cytologic
conversion within three months, and the remaining six patients showed this conversion at six months. The
duration of response ranged from 3 to 30 months; the duration of therapy was six months in most cases.
Two of the responders relapsed within the follow-up period
78
.
394
Schenkman and Lamm: Superficial Bladder Cancer Therapy TheScientificWorldJOURNAL (2004) 4 (S1), 387–399
Photofrin-Mediated Photodynamic Therapy (PDT)
Photofrin-Mediated Photodynamic Therapy (PDT) involves intravenous administration of
photosensitizers with subsequent in situ intravesical activation by use of whole bladder laser therapy
(WB-PDT) with visible light (630nm). Many compounds are being evaluated as potential
photosensitizers
79
; however, PHOTOFRIN, Porfimer Sodium (QLT Phototherapeutics, Inc., Vancouver,
BC, Canada) is the only photosensitizer which is approved by the U.S. Federal Drug Administration for
clinical use. PDT has been evaluated in therapy of recurrent superficial papillary TCC and refractory CIS,
in the prophylaxis of recurrent superficial TCC
80
. In studies involving 51 patients with TA and/or T1
TCC, CR occurred in 41%, while another 39% demonstrated PR following a single PDT treatment. For
papillary TCC, tumor size was a factor ñ CR was observed only in tumors less than 2 cm in diameter. In a
multicenter randomized trial involving 36 patients, preliminary findings on 24 patients indicate a
reduction in recurrence from 83-33% (net benefit of 50%) with a single prophylactic PDT treatment
following a complete transurethral resection of the bladder tumors
80
. The median time to tumor recurrence
increased from 3-13 months with the addition of a single adjuvant PDT treatment. Long-term data on
prevention of recurrence and progression are lacking.
An important application of PDT treatment is in the management of refractory/recurrent CIS. In a
single whole bladder, PDT treatment produced a complete response in 88% of the patients, and only 25%
of the patients experienced recurrence during follow-ups ranging from 3-55 months. Recently Nseyo et
al.
81
reported on a multicenter clinical trial involving 35 patients who received a single PDT treatment for
refractory CIS as an alternative to cystectomy. All patients had failed standard regimens of at least two
intravesical therapies including BCG. The authors showed that PDT treatment induced complete
responses in 52% of these highest risk patients, and spared 58% of the patients radical cystectomy.
The mechanisms of action of PDT include direct cytotoxicity mediated by singlet oxygen and
superoxide radicals; vascular endothelial damage with thrombosis and hypoxia; and intense local
inflammation associated with immune response. Consequently, PDT treatment induces symptoms of
cystitis (post-PDT syndrome) urinary frequency, urgency, nocturia, suprapubic pain, and bladder spasm.
The intensity of duration of these symptoms depend directly on PDT dose (light and PHOTOFRIN),
extent of detrusor damage from previous treatments, intensity of a acute inflammation, and CIS (which
enhances PHOTOFRIN retention). The most severe adverse reaction of PDT treatment is permanent
bladder contracture which has been reported in 4-24% of patients
80,81
. With proper patient education and
selection, the problem of PHOTOFRIN -induced skin photosensitivity has been minimal. However,
avoidance of direct exposure to sunlight is required up to six weeks after PHOTOFRIN injection.
Introduction of new photosensitizers and simplification of WB-PDT laser light delivery may lead to wide
clinical application of PDT in the management of bladder cancer.
CONCLUSION
Intravesical BCG remains the most effective therapy in the management and prophylaxis of superficial
TCC of the urinary bladder. BCG intravesical immunotherapy has improved tumor recurrence rates,
disease progression rates, and has prolonged survival of patients with this disease. Intravesical
chemotherapy, on the other hand, has reduced tumor recurrence rates but has had no positive impact on
disease progression or survival. Although results from newer intravesical therapies such as IFN, KLH,
bropirimine, and PDT are encouraging, to date they have only proven to be useful in the therapy and
prophylaxis of superficial TCC and long-term data on the prevention of recurrence, disease progression,
and survival are unknown.
395
Schenkman and Lamm: Superficial Bladder Cancer Therapy TheScientificWorldJOURNAL (2004) 4 (S1), 387–399
REFERENCES
1. Parker, S.L., Tony, T., Bolden, S., Wingo, P.A.: Cancer Statistics, 1997. CA Cancer J Clin. 1997; 47:5-27.
2. Cohen, S.M., Johansson, S.L.: Epidemiology and etiology of bladder cancer. Urol Clin North Amer. 1992; 19:421-
428.
3. Ro, J.Y., Staerkel, G.A., Ayala, A.G.: Cytologic and histologic features of superficial bladder cancer. (Review) J Urol
Clin North Amer. 1992; 19:435-449.
4. Lamm, D.L., Griffith, J.G.: Intravesical Therapy: Does it affect the natural history of bladder cancer? Semin Urol
1992; 10:39-44.
5. Althausen, A.F., Prout, G.R., Jr., Daly, J.J.: Noninvasive papillary carcinoma of the bladder associated with carcinoma
in situ. J Urol 1976; 116:575-580.
6. Lutzeyer, W., Rubben, H., Dahm, H.: Prognostic parameters in superficial bladder cancer: an analysis of 315 cases. J
Urol 1982; 127:250-252.
7. Heney, N.M., Ahmed, S., Flanagan, M.J., et al: Superficial bladder cancer: Progression and recurrence. J Urol 1983;
130:1083-1086.
8. Abel, P.D.: Follow-up of patients with "superficial" transinsulars cell carcinoma of the bladder: The case for a change
in policy. Br J Urol, 1993, 72:135-142.
9. Bostwick, D.G.: Natural history of bladder cancer. J Cell Biochem., 1992;161:31-38.
10. Lippenon, P.: The changing important of prognostic factors in bladder cancer during a long-term follow up, Eur J
Cancer, 1993, 29A:1261-1265.
11. Lamm, D.L.: Carcinoma in situ (review) Urol Clin North Amer. 1992; 19:499-508
12. Lamm, D.L., Blumenstein, B.A., Crawford, E.D., Montie, J.E., Scardino, P., Grossman, H.B., Stanisic, T.H., Smith,
H.A., Sullivan, J., Sarosdy, M.F., Crissman, J.D., and Coltman, C.A.: A randomized trial of intravesical doxorubicin
and immunotherapy with Bacille Calmette-Guerin for transitional cell carcinoma of the bladder. New Engl J Med.
1991, 325(17):1205-1209.
13. Walker, M.C., Masstera, J.R., Parris, C.N., et al: Intravesical chemotherapy in vitro studies on the relationship
between dose and cytotoxicity. Urol Res. 1986; 14-137-140.
14. Traynelis, C.L. and Lamm, D.L.: Current status of intravesical therapy for bladder cancer, In: Urology Annual Vol 8,
Stephen N. Rous (Ed.). W.W. Norton & Company, 1994, pp. 113-143.
15. Melekos, M.D.: Prophylaxis of superficial bladder cancer with a modified intravesical epirubicin treatment schedule.
Oncology, 1993, 50:450-455.
16. Oosterllinck, W., Kurth, K.H., Schroder, F., Bultinck, J., Hammond, B., Sylvester, R., and Members of the European
Organization for Research and Treatment of Cancer Genitourinary Group. A prospective European organization of
research and treatment of cancer genitourinary group randomized trial comparing transurethral resection followed by a
single intravesical instillation of epirubicin or water in a single state Ta, T1 papillary carcinoma of the bladder. J Urol,
1993; 149:749-752.
17. Pawinski, A., Bouffioux, C., Sylvester, R., et al: Meta-analysis of EORTC/MRC random mixed clinical trials for
prophylactic treatment of TA,T1 bladder cancer. J Urol, 1996; 155:492A (727)
18. Sekine, H., Fukui, I., Yamada, T., Ohwada, F., Yokokawa, M., Oshima, H.: Intravesical mitomycin-C and
doxorubicin sequential therapy for carcinoma in situ of the bladder. J Urol. 1994, 151:1:27-30.
19. Wientjes, M.G., Badalament, R.A., Wang, R.C., Hassan, F., Au, J.L.: Penetration of Mitomycin C in human bladder.
Cancer Res 1993, 53:3314-3320.
20. Davies, M.: Bacillus Calmette-Guerin as an antitumor agent. The interaction with cells of the mammalian immune
system. Biochem Biophys Acta. 1982;651:143.
21. DeBoer, E.C., DeJong, W.H., Steerenberg, P.A., et al,: Induction of urinary interleukin-1 (IL-1), IL-2, IL-6, and tumor
necrosis factor during intravesical immunotherapy with bacillus Calmette-Guerin in superficial bladder cancer. Cancer
Immunol Immunother 1992; 34:306-312.
22. McAveray, D.M., Gomella, L.G., Lattime, E.C.: Cytokin regulation of antitumor immunity following localized
immunotherapy of bladder cancer. (Abstract) Proceedings of the 85th Annual Meeting of the American Association
for Cancer Research; 1994 April 10-13; San Francisco, (CA):35:A2863.
23. Ratliff, T.L., Ritchey, J.K., Yaun, J.J., et al.: Role of CD4 and CD8 lymphocytes in the antitumor activity of BCG. J
Urol 1993; 150:1018-1023.
24. Sosnowski, J.T., Lamm, D.L.: Immunotherapy for bladder cancer. In Rous SN, ed. Urology Annual, East Norwalk,
Connecticut: Appleton & Lange; 1990;4:123-156.
25. Morales, A., Ottenhof, P.C.: Clinical application of a whole blood assay for human natural killer (NK) cell activity.
Cancer. 1983;52:667.
26. Conti, P., Reale, M., Nicolai, M., et al: Bacillus Calmette-Guerin potentiates monocytes responses to
lypopolsaccharide-induced tumor necrosis factor and interleukin-1, but notinterleukin-6 in bladder cancer patients.
Cancer Immunol Immunother 1994;38:365-271.
27. Ratliff, T.L., Gillen, D., Catalona, W.J.: Requirement of a thymus dependent immune response for BCG-mediated
antitumor activity. J Urol. 1987;137:155.
396
Schenkman and Lamm: Superficial Bladder Cancer Therapy TheScientificWorldJOURNAL (2004) 4 (S1), 387–399
28. Ratliff, T.L., Hudson, M.A., Catalona, W.J.: Strategy for improving therapy of superficial bladder cancer. World J
Urol. 1991;9:95.
29. Becich, M.J., Carroll, S., Ratliff, T.L.: Internalization of Bacille Calmette-Guerin by bladder tumor cells. J Urol.
1991;145:1316.
30. Kuroda, K., Brown, E.J., Telle, W.B., et al: Characterization of the internalization of bacillus Calmette-Guerin by
human bladder tumor cells, J Clin Invest 1993;91:69-76.
31. Morales, A., Nickel, J.C.: Immunotherapy of superficial bladder cancer with BCG. World J Urol 1986;3:209-214.
32. Garden, R.J., Liu, B.C.S., Redwood, S.M., et al.: Bacillus Calmette-Guerin abrogates in vitro invasion and motility of
human bladder tumor cells via fibronectin interaction. J Urol. 1992;148:900.
33. Schmidt, A.C., Bouic, P.J., Heyns, C.F., DeKock, M.L.: Peripheral blood lymphocyte response in patients with
superficial transitional cell carcinoma of the bladder treated with intravesical bacillus Calmette-Guerin - a useful
marker of response? Br J Urol 1993;71:179-182.
34. Winters, W.D., Lamm, D.L.: Antibody responses to bacillus Calmette-Guerin during immunotherapy in bladder
cancer patients. Cancer Res 1981;41:2672-2676.
35. Lamm, D.L., Reichart, D.F., Harris, S.C., Lucio, R.M.: Immunotherapy of murine transitional cell carcinoma, J Urol
128:1104, 1982.
36. Corti Ortiz, D., Rivera, Garay, P., Aviles Jasse, J., et al: (Prophylaxis of superficial bladder cancer with 1 mg of
intravesical BCG: comparison with other doses). (Spanish) Actas Urol Esp 1993; 17:239-242.
37. Ratliff, T.L., Kavoussi, L.R., Catalona, W.J.: Role of fibronetin in intravesical BCG therapy for superficial bladder
cancer. J Urol 188; 139:410-414.
38. Hudson, M.A., Yvan, J.J., Catalona, W.J., Ratliff, T.L.: Adverse effect of fibrin clot inhibitors on intravesical bacillus
Calmette-Guerin therapy for superficial bladder tumors. J Urol 1990; 144:1362-1364.
39. Rogerson, J.W.: Intravesical bacille Calmette-Guerin in the treatment of superficial transitional cell carcinoma of the
bladder. Br J Urol 1994; 73:655-658.
40. DeBoer, L.C., Steerenberg, P.A., Van der Meijden, P.M., et al: Impaired immune response by isoniazid treatment
during intravesical BCG treatment in the guinea pig. J Urol 1992; 148:1477-1582,
41. Stassar, M.J., Vegt, P.D., Steerenber, P.A., et al.: Effects of isoniazid (INH) on the BCG-induced local
immunotherapy response after intravesical BCG therapy for superficial bladder cancer. Urol Res 1994; 22:177-184.
42. Lamm, D.L.: (1992) Long-term results of intravesical therapy for superficial bladder cancer. In Lamm DL, ed. The
Urologic Clinics of North America, Philadelphia, PA: W.B. Saunders Co.; 19:573-580.
43. Nadler, R.B., Catalona, W.J., Hudson, M.A., Ratliff, T.L.: Durability of tumor-free response for intravesical bacillus
Calmette-Guerin therapy. J Urol 1994; 152(2 Pt 1):367-373.
44. Herr, H.W.: Transurethral resection and intravesical therapy of superficial bladder tumors. In Fair WR, ed. The
Urologic Clinics of North America, Philadelphia, PA: W.B. Saunders Co.; 1991; 18:525-528.
45. Lamm, D.L.: (1985) Bacillus Calmette-Guerin immunotherapy for bladder cancer. Journal of Urology. 134:40-47.
46. Herr, H.W., Pinsky, C.M., Whitmore, W.F., et al.: Experience with intravesical Bacillus Calmette-Guerin therapy of
superficial bladder tumors. Urology 1985; 25:119.
47. Herr, H.W., Pinsky, C.M., Whitmore, W.F., et al.: Long-term effect of intravesical Bacillus Calmette-Guerin on flat
carcinoma in situ of the bladder. J Urol 1986; 135:265.
48. Pagano, F., Bassi, P., Milani, C., et al.: A low dose bacillus Calmette-Guerin regimen in superficial bladder cancer
therapy: Is it effective?. J Urol. 1991;146:32.
49. Melekos, M.D., Chionis, H., Pantazakos, A., Fokaefs, E., Paranychianakis, G., and Dauaher, H.: Intravesical Bacillus
Calmette-Guerin Immunoprophylaxis Of Superficial Bladder Cancer: Results Of A Controlled Prospective Trial With
Modified Treatment Schedule. J Urol 1993;149:744.
50. Krege, S., Giani, G., Meyer, R., et al.: A randomized multicenter trial of adjuvant therapy in superficial bladder
cancer: transurethral resection only versus transurethral resection plus mitomycin versus transurethral resection plus
Bacillus Calmette-Guerin. J Urol 1996; 156: 962-6.
51. Rodriques Netto Junior, N., Lemos, G.C.: A comparison of treatment methods for prophylaxis of recurrent superficial
bladder tumors. J Urol 1983; 129:33-34.
52. Martinez-Pineiro, J.A., Leon, J.J., Martinez-Pineiro, L. Jr., Fiter, L., Mosteiro, J.A., Navarro, J., Garcia, Matres, M.J.,
Carcamo: Bacillus Calmette-Guerin versus Doxorubicin versus Thiotepa: A randomized prospective study in 202
patients with superficial bladder cancer. Journal of Urology 1990;143:502-506.
53. Debruyne, F.M.J., Van der Meijden, A.P., Goebers, A.D.H., et al.: BCG-RIVM versus mitomycin intravesical therapy
in superficial bladder cancer first results of a randomized prospective trial. Urology (suppl) 1988; 31:20-25.
54. Jauhianen, K., Rintala, E., Alfthan, O., and the Finnbladder Group: Immunotherapy (BCG) versus chemotherapy
(MMC) in intravesical treatment of superficial urinary bladder cancer. In deKernion JB, ed. Immunotherapy of
Urological Tumors, Edinburgh: Churchill Livingston, 1990, 13-60.
55. Rubben, H., Graf-Dobberstein, C., Ostwald, R., et al.: Prospective randomized study of adjuvant therapy after
complete resection of superficial bladder cancer; mitomycin C vs. BCG Connaught vs. TUR alone. In deKernion JB,
ed. Immunotherapy of Urological Tumors, Edinburgh: Churchill Livingstone; 1990;27-36.
397
Schenkman and Lamm: Superficial Bladder Cancer Therapy TheScientificWorldJOURNAL (2004) 4 (S1), 387–399
56. Witjes, J.A., Van der Meijden, A.P., Witjes, J.P., et al.: Randomized prospective study comparing intravesical
instillations of mitomycin-C, BCG-TICE, and BCG-RIVM in Pta-Pt1 tumors and primary carcinoma in situ of the
urinary bladder. Dutch South-East Cooperative Group. Eur J Cancer 1993;29A:1672-1676.
57. Lamm, D.L., Crawford, E.D., Blumenstein, B., et al.: SWOG 8795: A randomized comparison of bacillus Calmette-
Guerin and mitomycin-C prophylaxis in stage Ta and T1 transitional cell carcinoma of the bladder. Urol Oncol 1995;
1:119-126.
58. Huland, H., Otto, U.: Use of mitomycin as prophylaxis following endoscopic resection of superficial bladder cancer.
Urology 1985; 26(4 Suppl):32-35.
59. Melekos, M.D., Chionis, H.S., Paranychianakis, G.S., Dauaher, H.H.: Intravesical 4'-epi-doxorubicin (epirubicin)
versus bacillus Calmette-Guerin. A controlled prospective study on the prophylaxis of superficial bladder cancer.
Cancer 1993; 72:1749-1755.
60. Cookson, M.S., Sarosdy, M.F.: Management of stage T1 superficial bladder cancer with intravesical bacillus
Calmette-Guerin therapy. Journal of Urology. 1992;148:797-801.
61. Herr, H.W., Laudone, V.P., Badalament, R.A., et al.: Bacillus Calmette-Guerin therapy alters the progression of
superficial bladder cancer. J Clin Oncol 1988; 6:1450-1455.
62. Witjes, J.A., Fransen, M.P., Van der Meijden, A.P., et al.: use of maintenance intravesical bacillus Calmette-Guerin
(BCG), with or without intradermal BCG, in patients with recurrent superficial bladder cancer. Long-term follow-up
of a randomized phase 2 study. Urol Int 1993; 51:67-72.
63. Lamm, D.L.: BCG in perspective: advances in the treatment of superficial bladder cancer. Eur Urol 1995; 27 (Suppl
1):2-8.
64. Lamm, D.L., Riggs, D., Shriver, J., Van Gilder, P., Rach, J., DeHaven, J.: Megadose vitamins in bladder cancer: A
double-blind clinical trial. J Urol 1994;151:21-26.
65. Lamm D.L.: Complications of bacillus Calmette-Guerin immunotherapy. (Review) Urol Clin North Am 1992;
19:565-572.
66. Berry D.L., Blumenstein B.A., Magyary D.L., et al.: Local toxicity patterns associated with intravesical bacillus
Calmette-Guerin: A Southwest Oncology Group Study. Int J Urol 1995; 3:98-101.
67. Koukol, S.C., DeHaven, J.I., Riggs, D.R., Lamm, D.L.: Drug therapy of bacillus Calmette-Guerin sepsis. Urol Res
1995; 22:373-376.
68. Bowyer, L., Hall, R.R., Reading, J., March M.M.: The persistence of bacille Calmette-Guerin in the bladder after
intravesical treatment of bladder cancer. Br J Urol 1995; 75:199-192.
69. Kristjansson, M., Green, P., Manning, H.L., RD, et al.: Molecular confirmation of bacillus Calmette-Guerin as the
cause of pulmonary infection following urinary tract instillation. Clin Infect Dis 1993; 17:228-230.
70. Torti, F.M., Shortliffe, L.D., Williams, R.D., Pitts, W.C., Kempson, R.L., Ross, J.C., Palmer, J., Meyers, F., Ferrari,
M., Hannigan, J., Spiegel, R., McWhirter, K., and Freihia, F.: Alpha-Interferon in superficial bladder cancer: A
northern California oncology group study. J Clin Oncol 1988;6:476.
71. Glashan, R.: A randomized controlled study of intravesical a-2b-interferon in carcinoma in situ of the bladder. J Urol.
1990;144:658.
72. Jurincic, C.D., Engelmann, U., Gasch, J., et al.: Immunotherapy in bladder cancer with keyhole-limpet hemocyanin: A
randomized study. J Urol. 1988;139:723..
73. Flamm, J., Bucher, A., Holtl, W., et al.: Recurrent superficial transitional cell carcinoma of the bladder: Adjuvant
topical chemotherapy versus immunotherapy: A prospective randomized trial. J Urol. 1990;144:260.
74. Lamm, L.D., Morales, A., Grossman, H.B., et al.: Keyhole limpet hemocyanin (KLH) immunontherapy of papillary
and in situ transitional cell carcinoma of the bladder. A multicenter phase I-II clinical trial. (Abstract) J Urol 1996;
155:A1405.
75. Wishahi, M.M., Ismail, I.M., Rubben, H., Otto, T.: Keyhole-limpet hemocyanin immunotherapy in the bilharzial
bladder: a new treatment modality? Phase II trial: superficial bladder cancer. J Urol 1995; 153(3 Pt 2):926-928.
76. Lamm, D.L., DeHaven, J.I., Riggs, D.R., et al.: Keyhole limpet hemocyanin immunotherapy of murine bladder
cancer. Urol Res 1993; 21:33-37.
77. Sarosdy, M.F., Lowe, B.A., Schellhammer, P.F., et al.: Bropirimine immunotherapy of bladder CIS: positive phase II
results of an oral interferon inducer. (Abstract) Proc Annu Meet Am Soc Clin Oncol 1994; 13:A719.
78. Sarosdy, M.F., Lowe, B.A., Schellhammer, P.F., et al.: Oral bropirimine immunotherapy of carcinoma in situ of the
bladder: results of a phase II trial. Urology 1996; 48:21-27.
79. Ash, D.V., Brown, S.B.: New drugs and future developments in photodynamic therapy. Eur J Cancer 1993;29A:1781-
1783.
80. Nseyo, U.O.: Photodynamic Therapy. In Lamm DL, ed. The Urologic Clinics of North America, Philadelphia, PA:
W.B. Saunders Co.; 1992;19:591-599.
81. Nseyo, U.O., Crawford, E.D., Shumaker, B., et al.: Photodynamic therapy as an alternative to cystectomy in refractory
carcinoma in situ. (Abstract) Proceedings of the American Association of Cancer Research 86th Annual Meeting;
1995 March 18-22; Toronto, Canada:36:A1856.
398
Schenkman and Lamm: Superficial Bladder Cancer Therapy TheScientificWorldJOURNAL (2004) 4 (S1), 387–399
This article should be referenced as follows:
Schenkman, E. and Lamm, D.L. (2004) Superficial bladder cancer therapy. TheScientificWorldJOURNAL 4 (S1), 387–399.
Handling Editor:
Anthony Atala, Principle Editor
for Urology — a domain of TheScientificWorldJOURNAL.
399
... BCG immunotherapy remains the most effective treatment and prophylaxis in superficial transitional cell carcinoma (TCC) and reduces tumor relapse, disease progression and mortality. Interferons, KLH (Keyhole-limpet Hemocyanin), Bropirimine and Photofrin-PDT are being studied in the treatment of TCC and the results obtained are encouraging [43]. It is worth discussing that TGF-β1 is a multifunctional cytokine and is synthesized by nearly all cells and affects many physiological processes; its particular cell-specific activity depends on the cell environment and is stored as a large latent complex in the extracellular matrix (ECM), waiting to be activated and to establish a mechanical checkpoint in the progression of tissue repair. ...
... Some authors have reported that growing tumor cells can also be the source of transforming growth factor, entailing a specific immunosuppression observed in patients with malignancies; others suggest that some members of TGF-β1 superfamily, such as activin A, might be associated with tumor aggressiveness [42]. Some authors describe a local TGF-β1 inhibition by PDT, preventing vascular smooth muscle injury [43]. Adamek and coworkers have observed a significant decrease of TGF-β1 serum level after the topical use of 10% delta-aminolevunic acid (an endogenous non-proteinogenic amino acid) and showed that the immune system response in patients undergoing PDT due to basal cell carcinoma (BCC) are promising [44]. ...
Article
Full-text available
Photodynamic therapy (PDT) is a non-invasive and modern form of therapy. It is used in the treatment of non-oncological diseases and more and more often in the treatment of various types of neoplasms in various locations including bladder cancer. The PDT method consists of local or systemic application of a photosensitizer, i.e., a photosensitive compound that accumulates in pathological tissue. Light of appropriate wavelength is absorbed by the photosensitizer molecules, which in turn transfers energy to oxygen or initiates radical processes that leads to selective destruction of diseased cells. The technique enables the selective destruction of malignant cells, as the photocytotoxicity reactions induced by the photosensitizer take place strictly within the pathological tissue. PDT is known to be well tolerated in a clinical setting in patients. In cited papers herein no new safety issues were identified. The development of anti-cancer PDT therapies has greatly accelerated over the last decade. There was no evidence of increased or cumulative toxic effects with each PDT treatment. Many modifications have been made to enhance the effects. Clinically, bladder cancer remains one of the deadliest urological diseases of the urinary system. The subject of this review is the anti-cancer use of PDT, its benefits and possible modifications that may lead to more effective treatments for bladder cancer. Bladder cancer, if localized, would seem to be a good candidate for PDT therapy since this does not involve the toxicity of systemic chemotherapy and can spare normal tissues from damage if properly carried out. It is clear that PDT deserves more investment in clinical research, especially for plant-based photosensitizers. Natural PS isolated from plants and other biological sources can be considered a green approach to PDT in cancer therapy. Currently, PDT is widely used in the treatment of skin cancer, but numerous studies show the advantages of related therapeutic strategies that can help eliminate various types of cancer, including bladder cancer. PDT for bladder cancer in which photosensitizer is locally activated and generates cytotoxic reactive oxygen species and causing cell death, is a modern treatment. Moreover, PDT is an innovative technique in oncologic urology.
... For a clinically correct diagnosis and course of treatment, it is essential to identify molecular markers that are possible therapeutic targets and prognostic indicators of bladder cancer. The management of this condition depends on having a thorough understanding of the prognostic factors and natural history of bladder cancer (Schenkman & Lamm, 2004). Antimetabolite mitomycin is utilized as a chemotherapeutic medication for various malignancies as well as an addition to ab externo (outside approach) eye operations for the treatment of glaucoma. ...
... Effective PDT required enough O 2 to succeed photooxidation progression (Price et al., 2013). PDT has been primarily applied as a local therapy for malignancies such as skin cancer (Alter et al., 2015) and superficial bladder cancer (Emmanuel and Donald 2004). However, it has recently become more widely accepted as a treatment option for early gastric, esophageal, and lung cancer (Aiwu, 2016;Oinuma et al., 2016;Harris et al., 2017). ...
Article
Full-text available
Indocyanine green (ICG) is a nontoxic registered photosensitizer used as a diagnostic tool and for photodynamic therapy (PDT). Hypoxia is one the main factors affecting PDT efficacy. Perfluorodecalin emulsion (Perftoran ® ) is a known oxygen carrier . This study investigated the effect of Perftoran ® on ICG/PDT efficacy in presence and absence of Perftoran ® via evaluation of phototoxicity by MTT; hypoxia estimation by pimonidazole, HIF-1 α / β by ELISA, and 17 miRNAs (tumor suppressors, oncomiRs, and hypoxamiRs) were analyzed by qPCR. Compared to ICG/PDT, Perftoran ® /ICG/PDT led to higher photocytotoxicity, inhibited pimonidazole hypoxia adducts, inhibited HIF-1 α / β concentrations, induced the expression of tumor-suppressing miRNAs let-7b/d/f/g, and strongly inhibited the pro-hypoxia miRNA let-7i. Additionally, Perftoran ® /ICG/PDT suppressed the expression of the oncomiRs miR-155, miR-30c, and miR-181a and the hypoxamiRs miR-210 and miR-21 compared to ICG/PDT. In conclusion, Perftoran ® induced the phototoxicity of ICG/PDT and inhibited ICG/PDT-hypoxia via suppressing HIF- α / β , miR-210, miR-21, let-7i, miR-15a, miR-30c, and miR-181a and by inducing the expression of let-7d/f and miR-15b.
... Furthermore, heat is generated by the reaction, and this thermal effect also contributes to the tumor suppressing effect [4]. PDT is mainly applied as a topical therapy for malignant tumors such as skin cancer [7] and superficial bladder cancer [8]. In addition, PDT has recently been widely accepted as a treatment option for early gastric, esophageal, and lung cancers [9]. ...
Article
Full-text available
BACKGROUND: The diagnostic use and therapeutic effect of near infrared fluorescence (NIF) imaging and photodynamic therapy (PDT) was investigated for gallbladder cancer using indocyanine green (ICG)-lactosomes. RESULTS: PDT was toxic for NOZ cells treated with ICG-lactosomes. Fluorescence intensity in the tumor region of mice administered ICG-lactosomes, but not ICG alone, was higher than the healthy contralateral region ≥24 hours after injection. PDT exerted immediate and continuous phototoxic effects in NOZ implanted mice injected with ICG-lactosomes. Enhanced antitumor effects were observed in the twice irradiated group compared with the once irradiated group. METHOD: ICG or ICG-lactosomes were added to the human gallbladder cancer cell line NOZ followed by PDT and cell viability was measured. Mass spectrometry of ICG and ICG-lactosomes was performed after PDT. ICG or ICG-lactosomes were intravenously administered to BALB/c nude mice implanted subcutaneously with NOZ cells and fluorescence was evaluated by NIF imaging. Implanted tumors underwent PDT and antitumor effects were analyzed after performing irradiation once or twice in ICG-lactosome groups. CONCLUSIONS: ICG-lactosomes accumulated in xenograft tumors and PDT had an antitumor effect on these malignant tumors. NIF imaging with ICG-lactosomes and PDT may be useful diagnostic and/or therapeutic agents for gallbladder cancer.
... Jurincic и съавт. съобщават, че 95% от паци- ентите, лекувани с KLH, са имали пълен отговор към терапията, в сравнение с 69% от пациентите лекувани с Mitomycin C. Този клиничен отговор на KLH е сравним с клиничния отговор към BCG, макар и с по-малко странични ефекти [73,74]. В проучване, което включва 101 пациенти, само три- ма са имали леко повишаване на температурата след началното интрадермално инжектиране, като не е наблюдавана системна токсичност или други неблагоприятни странични ефекти [65]. ...
Article
Full-text available
Summary. Bladder cancer is the most common malignancy of the urinary system. The main reasons for its appearance are urinary tract infections and arylamines which are in direct contact with the wall of the bladder. The treatment of bladder cancer depends mainly on the histological characteristics of the tumor, invasion depth in the bladder wall and the cancer recurrence. The bladder cancer treatment is surgical removal of the tumor, combined with radiotherapy, chemotherapy or immunotherapy. In the treatment of superfi cial non-invasive bladder cancer, chemotherapy is used prophylactically as adjuvant intravesical therapy after endoscopic resection of the tumor. The most frequently used chemotherapeutic drugs are mitomycin, epirubicin, doxorubicin, and thiotepa. More than half of the patients with invasive bladder cancer die due to distant metastases. These metastases develop early in the process of tumor invasion. A combination of local treatments and systemic chemotherapy is used to reduce the number and depth of the micrometastases. The reduction of the size of the primary tumor is achieved by neoadjuvant chemotherapy with methotrexate, vinblastine, adriamycin and cisplatin. The main method of treatment of the patients with advanced metastatic bladder cancer is chemotherapy. Two modes of chemotherapy are applied: M-VAC (Methotrexate, Vinblastine, Adriamycin, Cisplatin) and CMV (cisplatin, methotrexate, vinblastine). Another approach used in the treatment of the bladder cancer is immunotherapy, which stimulate the immune system of the patient to fi ght cancer. For example intravesical administration of BCG (Bacillus Calmette-Guérin) is used as an immunostimulant in treatment of bladder carcinoma in situ (CIS), This approach decreases the disability as a consequence of the therapeutic cystectomy.
Article
Full-text available
Bladder cancer is first cancer for which PDT was clinically approved in 1993. Unfortunately, it was unsuccessful due to side effects like bladder contraction. Here we summarized the recent progress of PDT for bladder cancers, focusing on photosensitizers and formulations. General strategies to minimize side effects are intravesical administration of photosensitizers, use of targeting strategies for photosensitizers, and better control of light. Non‐muscle invasive bladder cancers are more suitable for PDT than muscle‐invasive and metastatic bladder cancers. In 2010, the FDA approved blue light cystoscopy, using PpIX fluorescence, for photodynamic diagnosis of non‐muscle invasive bladder cancer. PpIX produced from HAL was also used in PDT but wasn't successful due to low therapeutic efficacy. To enhance the efficacy of PpIX‐PDT, we have been working on combining it with singlet‐oxygen‐activatable prodrugs. The use of these prodrugs increases the therapeutic efficacy of the PpIX‐PDT. It also improves tumor selectivity of the prodrugs due to the preferential formation of PpIX in cancer cells resulting in decreased off‐target toxicity. Future challenges include improving prodrugs and light delivery across the bladder barrier to deeper tumor tissue and generating an effective therapeutic response in an in vivo setting without causing collateral damage to bladder function.
Article
Sosnovsky's hogweed, Heracleum sosnowskyi has a high photosensitizing ability. Although Sosnovsky's hogweed is known as a poisonous plant, its chemical composition and phototoxicity are poorly studied. We analyzed the chemical composition of the Sosnovsky's hogweed juice that grew in natural conditions. It was found that the content of 8-methoxypsoralen in the juice is 1332.7 mg/L, and that of 5-methoxypsoralen is 34.2 mg/L. We have developed and analyzed liposomes containing furanocoumarins of Sosnovsky's hogweed juice and studied their photocytotoxicity in L929 mouse fibroblast cell culture. It was found that liposomes containing furanocoumarins of Sosnovsky's hogweed juice are more toxic for L929 cells in comparison with liposomal forms of pure substances 8-methoxypsoralen and 5-methoxypsoralen. It was found that when exposed to UV radiation at 365 nm at a dose of 22.2 J/cm², the liposomal form of furanocoumarins Sosnovsky's hogweed is 3 times more toxic to L929 cells than in the dark. It was found that the photocytotoxic effect of liposomal furanocoumarins Sosnovsky's hogweed is a strongly stimulation of apoptosis.The data obtained suggest that the raw material of Sosnovsky's hogweed claims to be a source of furanocoumarins, and the liposomal form, given the hydrophobic properties of furanocoumarins, is very suitable for creating a drug for photodynamic therapy.
Article
Full-text available
This article presents the results of a clinical study that examined the diagnostic efficacy of fluorescent diagnostics (FD) of non-muscularinvasive bladder cancer using a photosensitizer of FD of malignant neoplasms – 5-aminolevulinic acid hexyl ester (5-ALA HE) compared with standard cystoscopy. The study involved 110 patients. The study began with intravesical administration of 50 ml of 0.2% solution of 5-ALA HE, the exposure time was 1 hour, after which the drug was removed from the organ. During the next hour, the mucous membranes were examined in two cystoscopy modes, followed by a standard transurethral resection of all urothelium sites with suspicion for tumor lesion based on white light and visible red fluorescence, and a control blind biopsy from the visually unchanged and non-fluorescent mucous tissue in each patient. The results of the study indicate the high effectiveness of the developed FD methodology with 5-ALA HE in detecting non-muscularinvasive bladder cancer during intravesical administration of the drug, due to selective accumulation of hexasens-induced PPIX in the tumor tissue compared with healthy mucosa. Compared with the results of standard cystoscopy, fluorescence diagnostics significantly increased diagnostic sensitivity by 24.4% (from 75.1% to 99.5%), diagnostic accuracy – by 15.8% (from 82.4% to 98.2%) and a negative predictive value – by 33.2% (from 65.8% to 99%) (p≤0.05). Additionally, a total of 37 (33.6%) patients was found to have 63 foci of fluorescence with a diameter of 2.5 to 3.0 mm. 59 of these were morphologically confirmed to contain cancer cells.
Article
Most bladder cancers present as a superficial disease, confined to the bladder mucosa or submucosal layer, without muscle invasion. Most superficial tumors have a propensity for recurrence after transurethral resection; some have a high risk for progression to muscle invasion. The treatment aim in superficial bladder cancer with intravesical therapy is three-fold: (1) eradicate existing disease, (2) prevention of recurrence, (3) prevention of tumor progression. The prognostic factors (tumor stage, grade, size, number and recurrence pattern) allow the stratification of tumors in different risk groups to plan treatment. Studies on pharmacokinetics have proved the efficacy of optimized drug delivery. Comparing resection with and without intravesical chemotherapy, a short-term reduction, approximately 15%, in tumor recurrence with chemotherapy can be obtained, but no effect on progression was proven. No agent has proved to be more effective than the others. A single instillation of chemotherapy immediately after transurethral resection has proven to be effective, but the role of maintenance therapy is controversial. Immunotherapy, in the form of Bacillus Calmette-Guerin, is generally shown to be more effective than chemotherapy, even if the results in comparison to mitomycin C do not result conclusive. Several new approaches are being explored to improve the efficacy of this therapy.
Article
Bacillus Calmette-Guerin (BCG) intravesical therapy represents a major advance in the treatment of superficial transitional cell carcinoma of the bladder. To date, however, the optimal treatment schedule must be defined and the toxicity related to the treatment is significant. The preliminary results of a randomized ongoing study performed to evaluate the effectiveness and relative toxicity of a low dose (75 mg.) BCG regimen in the treatment of superficial bladder cancer therapy are reported. A total of 126 patients (70 for prophylaxis of recurrent stages Ta and T1 papillary tumors and 56 for treatment of carcinoma in situ or with microinfiltration of the subepithelial connective tissue) underwent a 6-week course of 75 mg. BCG (Pasteur vaccine). An additional course was given in patients who failed to respond to the induction course. Maintenance therapy was administered in complete responders monthly for 1 year and then quarterly for 1 year. The prophylaxis group (transurethral resection plus BCG) was randomized versus transurethral resection alone (63 patients, control group). A complete response in the prophylaxis, control and therapy groups was observed in 74, 17 and 57% of the patients, respectively, while 4, 17 and 12.5%, respectively, experienced tumor progression. The additional course of therapy increased the response rate. On the contrary, previous unsuccessful intravesical chemotherapy did not affect the response rate. In regard to toxicity, irritative disturbances (27%) and fever (17%) appeared to be significantly decreased compared with the rates reported in the literature. No major complications were experienced. In conclusion, a low dose (75 mg.) Pasteur strain BCG regimen was effective as prophylaxis against recurrent superficial papillary tumors and as treatment of carcinoma in situ or with microinfiltration of the subepithelial connective tissue. Toxicity related to the treatment appeared to be low.
Article
Although intravesical bacillus Calmette-Guerin therapy has proved to be efficacious in the treatment and prophylaxis against tumor recurrence of superficial bladder tumors, its mechanism of action has not been fully elucidated. Previous work has suggested that bacillus Calmette-Guerin organisms attach to the matrix protein, fibronectin, during fibrin clot formation at sites of urothelial disruption and that this attachment was required for the antitumor effect of bacillus Calmette-Guerin to be expressed. Furthermore, drugs inhibiting clot formation were found to abrogate the antitumor effect of intravesical bacillus Calmette-Guerin therapy in a murine bladder tumor model.
Article
The tumors in 249 patients presenting initially with stages Ta and T1 bladder cancer were analyzed for tumor progression and recurrence. Only transurethral resection and/or fulguration was used before the first recurrence. Patients who received intravesical chemotherapy after the first tumor recurrence were excluded from an analysis of progression. Progression according to stages Ta and T1, and grades I, II and III was 4, 30, 2, 11 and 45 per cent, respectively. All differences were statistically significant. Progression also correlated with nontumor dysplasia and size. High tumor grade, lamina propria invasion, atypia elsewhere in the bladder, positive urinary cytology, tumor multiplicity and large tumors were associated with shorter intervals free of disease.
Article
The effectiveness of BCG in preventing disease progression in patients with superficial bladder cancer is evaluated. Long-term follow-up of high-risk patients treated in a previously reported randomized control trial of intravesical plus percutaneous BCG shows that progression occurred in 41/43 (95%) of control and 23/43 (53%) of BCG-treated patients. Muscle invasive and/or metastatic disease occurred with equal frequency in the two groups, but was significantly delayed by BCG treatment (P = .012). Cystectomies were required in 18/43 (42%) control and 11/43 (26%) BCG-treated patients. Median time to cystectomy was 8 months for control v 24 months for BCG-treated patients. Based on initial treatment, survival was improved by BCG therapy (P = .032) (median follow-up 6 years). These results suggest that in high-risk patients intravesical BCG can delay disease progression, prolong the period of bladder preservation, and increase overall survival.
Article
A randomized multicenter trial was done to compare transurethral resection only to transurethral resection plus adjuvant mitomycin C and bacillus Calmette Guerin (BCG) instillation for treatment of superficial bladder cancer (stage pTa/1 grades 1 to 3 except primary stage pTa grade 1). Included in the study were 337 patients with superficial stage pTa/1 grades 1 to 3 bladder cancer except primary stage pTa grade 1 tumors. One group underwent transurethral resection alone. Mitomycin C (20 mg./50 ml. sodium chloride) was given every 2 weeks during year 1 and once a month during year 2. BCG (120 mg/50 ml. sodium chloride was instilled once a week for 6 weeks and once a month for 4 months. At a median followup of 20.2 months, a decrease in recurrence rate was noted for both drug instillations compared to transurethral resection only. The relative risk of recurrence was 0.508 after mitomycin C and 0.618 after BCG instillation compared to transurethral resection alone. There was no significant difference between the mitomycin C and BCG instillations. The progression rate was comparable in all 3 therapy groups, with an estimated common progression rate of 4.22% per year. Side effects occurred most frequently during or after BCG instillation, most often consisting of cystitis. One patient required cystectomy because of ulcerating cystitis and a prostatic abscess subsequent to unsuccessful tuberculostatic therapy. There were no systemic complications. Our study showed a positive effect of adjuvant chemotherapy and immunotherapy on decreasing tumor recurrence rate. No influence was observed concerning progression rate, which was low overall.
Article
PurposeA randomized multicenter trial was done to compare transurethral resection only to transurethral resection plus adjuvant mitomycin C and bacillus Calmette Guerin (BCG) instillation for treatment of superficial bladder cancer (stage pTa/1 grades 1 to 3 except primary stage pTa grade 1).Materials and Methods Included in the study were 337 patients with superficial stage pTa/1 grades 1 to 3 bladder cancer except primary stage pTa grade 1 tumors. One group underwent transurethral resection alone. Mitomycin C (20 mg./50 ml. sodium chloride) was given every 2 weeks during year 1 and once a month during year 2. BCG (120 mg./50 ml. sodium chloride) was instilled once a week for 6 weeks and once a month for 4 months.ResultsAt a median followup of 20.2 months, a decrease in recurrence rate was noted for both drug instillations compared to transurethral resection only. The relative risk of recurrence was 0.508 after mitomycin C and 0.618 after BCG instillation compared to transurethral resection alone. There was no significant difference between the mitomycin C and BCG instillations. The progression rate was comparable in all 3 therapy groups, with an estimated common progression rate of 4.22 percent per year. Side effects occurred most frequently during or after BCG instillation, most often consisting of cystitis. One patient required cystectomy because of ulcerating cystitis and a prostatic abscess subsequent to unsuccessful tuberculostatic therapy. There were no systemic complications.Conclusions Our study showed a positive effect of adjuvant chemotheraphy and immunotherapy on decreasing tumor recurrence rate. No influence was observed concerning progression rate, which was low overall.
Article
Bacillus Calmette-Gurin (BCG) has achieved an important role in the prophylaxis and treatment of superficial bladder cancer. It has been found effective in the prevention of recurrences after endoscopic surgery as well as in the treatment of residual tumors. The most dramatic responses have been consistently demonstrated in the treatment of carcinoma in situ. Recent experience suggests that prolongation of treatment beyond the 6 weeks advocated in early protocols, significantly increases the anti-tumor activity of BCG. The side effects of intravesical administration of the vaccine are, in the vast majority of patients, well-tolerated, minimal and self-limiting. No permanent functional or structural damage of the bladder has been observed. The effectiveness of BCG is primarily dependent on the strain used as well as the dose, duration and frequency of its administration.